Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Angew Chem Int Ed Engl ; : e202408370, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39196601

RESUMEN

Native proteomics measures endogenous proteoforms and protein complexes under a near physiological condition using native mass spectrometry (nMS) coupled with liquid-phase separations. Native proteomics should provide the most accurate bird's-eye view of proteome dynamics within cells, which is fundamental for understanding almost all biological processes. nMS has been widely employed to characterize well-purified protein complexes. However, there are only very few trials of utilizing nMS to measure proteoforms and protein complexes in a complex sample (i.e., a whole cell lysate). Here, we pioneer the native proteomics measurement of large proteoforms or protein complexes up to 400 kDa from a complex proteome via online coupling of native capillary zone electrophoresis (nCZE) to an ultra-high mass range (UHMR) Orbitrap mass spectrometer. The nCZE-MS technique enabled the measurement of a 115-kDa standard protein complex while consuming only about 0.1 ng of protein material. nCZE-MS analysis of an E.coli cell lysate detected 72 proteoforms or protein complexes in a mass range of 30-400 kDa in a single run while consuming only 50-ng protein material. The mass distribution of detected proteoforms or protein complexes agreed well with that from mass photometry measurement. This work represents a technical breakthrough in native proteomics for measuring complex proteomes.

2.
Foods ; 13(16)2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39200451

RESUMEN

This study investigated the changes in the aroma of jasmine tea during storage. Solid-phase micro-extraction (SPME)-gas chromatography (GC)-mass spectrometry (MS) and stir bar sorptive extraction (SBSE)-GC-MS were combined to detect all volatile compounds. GC-olfactometry (GC-O), odor activity value (OAV), and p-value were employed to analyze and identify the key aroma compounds in six jasmine tea samples stored for different durations. Nine key aroma compounds were discovered, namely (Z)-3-hexen-1-yl acetate, methyl anthranilate, methyl salicylate, trans-ß-ionone, linalool, geraniol, (Z)-4-heptenal, benzoic acid methyl ester, and benzoic acid ethyl ester. The importance of these compounds was confirmed through the aroma addition experiment. Correlation analysis showed that (Z)-4-heptenal might be the main reason for the increase in the stale aroma of jasmine tea. Through sensory evaluation and specific experimental analysis, it can be concluded that jasmine tea had the best aroma after 3 years of storage, and too long a storage time may cause the overall aroma of the tea to weaken and produce an undesirable odor. The findings can provide a reference for the change in aroma during the storage of jasmine tea and provide the best storage time (3 years) in terms of jasmine tea aroma.

3.
J Transl Med ; 22(1): 538, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844946

RESUMEN

Apalutamide, a novel endocrine therapy agent, has been shown to significantly improve the prognosis of patients with metastatic hormone-sensitive prostate cancer (mHSPC). However, resistance to apalutamide has also been reported, and the underlying mechanism for this response has yet to be clearly elucidated. First, this study established apalutamide-resistant prostate cancer (PCa) cells, and confirmed that apalutamide activated the release of calcium ions (Ca2+) and endoplasmic reticulum stress (ERS) to enhance autophagy. Second, RNA sequencing, western blotting, and immunohistochemistry revealed significantly decreased Calpain 2 (CAPN2) expression in the apalutamide-resistant PCa cells and tissues. Furthermore, immunofluorescence and transmission electron microscopy (TEM) showed that CAPN2 promoted apalutamide resistance by activating protective autophagy. CAPN2 promoted autophagy by reducing Forkhead Box O1 (FOXO1) degradation while increasing nuclear translocation via nucleoplasmic protein isolation and immunofluorescence. In addition, FOXO1 promoted protective autophagy through the transcriptional regulation of autophagy-related gene 5 (ATG5). Furthermore, a dual-fluorescence assay confirmed that transcription factor 3 (ATF3) stimulation promoted CAPN2-mediated autophagy activation via transcriptional regulation. In summary, CAPN2 activated protective autophagy by inhibiting FOXO1 degradation and promoting its nuclear translocation via transcriptional ATG5 regulation. ATF3 activation and transcriptional CAPN2 regulation jointly promoted this bioeffect. Thus, our findings have not only revealed the mechanism underlying apalutamide resistance, but also provided a promising new target for the treatment of metastatic PCa.


Asunto(s)
Autofagia , Calpaína , Resistencia a Antineoplásicos , Metástasis de la Neoplasia , Neoplasias de la Próstata , Tiohidantoínas , Humanos , Masculino , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Calpaína/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Tiohidantoínas/farmacología , Tiohidantoínas/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Calcio/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Animales
4.
bioRxiv ; 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-38712154

RESUMEN

Native proteomics measures endogenous proteoforms and protein complexes under a near physiological condition using native mass spectrometry (nMS) coupled with liquid-phase separations. Native proteomics should provide the most accurate bird's-eye view of proteome dynamics within cells, which is fundamental for understanding almost all biological processes. nMS has been widely employed to characterize well-purified protein complexes. However, there are only very few trials of utilizing nMS to measure proteoforms and protein complexes in a complex sample (i.e., a whole cell lysate). Here, we pioneer the native proteomics measurement of large proteoforms or protein complexes up to 400 kDa from a complex proteome via online coupling of native capillary zone electrophoresis (nCZE) to an ultra-high mass range (UHMR) Orbitrap mass spectrometer. The nCZE-MS technique enabled the measurement of a 115-kDa standard protein complex while consuming only about 0.1 ng of protein material. nCZE-MS analysis of an E . coli cell lysate detected 72 proteoforms or protein complexes in a mass range of 30-400 kDa in a single run while consuming only 50-ng protein material. The mass distribution of detected proteoforms or protein complexes agreed well with that from mass photometry measurement. This work represents a technical breakthrough in native proteomics for measuring complex proteomes.

5.
Rev Sci Instrum ; 95(5)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38690983

RESUMEN

The power transformer is the core equipment of the power system, a sudden failure of which will seriously endanger the safety of the power system. In recent years, artificial intelligence techniques have been applied to the dissolved gas analysis evaluation of power transformers to improve the accuracy and efficiency of power transformer fault diagnosis. However, most of the artificial intelligence techniques are data-driven algorithms whose performance decreases when the data are limited or significantly imbalanced. In this paper, we propose an active learning framework for power transformer dissolved gas analysis, in which the model can be dynamically trained based on the characteristics of the data and the training process. In addition, this paper also improves the original active learning spatial search strategy and uses the product of sample feature differences instead of the original sum of differences as a measure of sample difference. Compared to passive learning algorithms, the novel approach could significantly reduce the data labeling effort while improving prediction accuracy.

6.
J Mol Biol ; 436(11): 168590, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38663547

RESUMEN

Redß is a protein from bacteriophage λ that binds to single-stranded DNA (ssDNA) to promote the annealing of complementary strands. Together with λ-exonuclease (λ-exo), Redß is part of a two-component DNA recombination system involved in multiple aspects of genome maintenance. The proteins have been exploited in powerful methods for bacterial genome engineering in which Redß can anneal an electroporated oligonucleotide to a complementary target site at the lagging strand of a replication fork. Successful annealing in vivo requires the interaction of Redß with E. coli single-stranded DNA-binding protein (SSB), which coats the ssDNA at the lagging strand to coordinate access of numerous replication proteins. Previous mutational analysis revealed that the interaction between Redß and SSB involves the C-terminal domain (CTD) of Redß and the C-terminal tail of SSB (SSB-Ct), the site for binding of numerous host proteins. Here, we have determined the x-ray crystal structure of Redß CTD in complex with a peptide corresponding to the last nine residues of SSB (MDFDDDIPF). Formation of the complex is predominantly mediated by hydrophobic interactions between two phenylalanine side chains of SSB (Phe-171 and Phe-177) and an apolar groove on the CTD, combined with electrostatic interactions between the C-terminal carboxylate of SSB and Lys-214 of the CTD. Mutation of any of these residues to alanine significantly disrupts the interaction of full-length Redß and SSB proteins. Structural knowledge of this interaction will help to expand the utility of Redß-mediated recombination to a wider range of bacterial hosts for applications in synthetic biology.


Asunto(s)
Bacteriófago lambda , ADN de Cadena Simple , Proteínas de Unión al ADN , Proteínas de Escherichia coli , Escherichia coli , Proteínas Virales , Bacteriófago lambda/genética , Bacteriófago lambda/metabolismo , Sitios de Unión , Cristalografía por Rayos X , ADN de Cadena Simple/metabolismo , ADN de Cadena Simple/química , ADN de Cadena Simple/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Escherichia coli/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Modelos Moleculares , Unión Proteica , Conformación Proteica , Proteínas Virales/metabolismo , Proteínas Virales/química , Proteínas Virales/genética
7.
Food Chem ; 451: 139458, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38670017

RESUMEN

Ripening refers to the process of chemical change during the refinement of Keemun black tea (KBT) and is crucial in the formation of Keemun Congou black tea's quality. In this study, the aroma composition of KBT during the ripening was analyzed. Sensomics indicated that ripening strengthened the coconut and fatty aroma of KBT and contributed to the decrease of green aroma substances, resulting in a shift of the overall aroma type of KBT to an integrated aroma profile, which was consistent with sensory evaluation. Changes in fatty acid content and the results of in vitro addition simulation tests confirmed that heat causes highly degradation of fatty acids into fatty aroma volatiles, which is a key driver of the formation of "Keemun aroma" quality. This study revealed the mechanism behind the formation of KBT's integrated "Keemun aroma" quality and the mode of thermal degradation of major fatty acids.


Asunto(s)
Ácidos Grasos , Calor , Odorantes , Compuestos Orgánicos Volátiles , Odorantes/análisis , Ácidos Grasos/metabolismo , Ácidos Grasos/química , Humanos , Compuestos Orgánicos Volátiles/química , Compuestos Orgánicos Volátiles/metabolismo , Compuestos Orgánicos Volátiles/análisis , Té/química , Té/metabolismo , Camellia sinensis/química , Camellia sinensis/metabolismo , Camellia sinensis/crecimiento & desarrollo , Aromatizantes/química , Aromatizantes/metabolismo , Manipulación de Alimentos
8.
iScience ; 26(1): 105723, 2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36590169

RESUMEN

Lipid metabolism is extensively reprogrammed in pancreatic ductal adenocarcinoma (PDAC). Stearoyl-coenzyme A desaturase (SCD) is a critical lipid regulator that was unexplored in PDAC. Here, we characterized the existence of cancer-associated fibroblasts (CAFs) with high SCD expression, and revealed them as an unfavorable prognostic factor. Therefore, primary CAFs and pancreatic cancer cells were harvested and genetically labeled. The mixture of CAFs and cancer cells were co-injected into scd-/-; prkdc-/-, or hIGF1/INS-expressing zebrafish to generate patient-derived xenograft models (zPDX). The models were aligned in 3D-printed chips for semi-automatic drug administration and high-throughput scanning. The results showed that chaperoning of the SCD-high CAFs significantly improved the drug resistance of pancreatic cancer cells against gemcitabine and cisplatin, while the administration of SCD inhibitors neutralized the protective effect. Our studies revealed the prognostic and therapeutic value of stromal SCD in PDAC, and proposed the application of zPDX model chips for drug testing.

9.
Genes (Basel) ; 13(9)2022 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-36140756

RESUMEN

Liver metastases are common in pancreatic neuroendocrine tumors (PanNETs) patients and they are considered a poor prognostic marker. This study aims to analyze the spatiotemporal patterns of genomic variations between primary and metastatic tumors, and to identify the key related biomolecular pathways. We performed next-generation sequencing on paired tissue specimens of primary PanNETs (n = 11) and liver metastases (n = 12). Low genomic heterogeneity between primary PanNETs and liver metastases was observed. Genomic analysis provided evidence that polyclonal seeding is a prevalent event during metastatic progression, and may be associated with the progression-free survival. Besides this, copy number variations of BRCA1/BRCA2 seem to be associated with better prognosis. Pathways analysis showed that pathways in cancer, DNA repair, and cell cycle regulation-related pathways were significantly enriched in primary PanNETs and liver metastases. The study has shown a high concordance of gene mutations between the primary tumor and its metastases and the shared gene mutations may occur during oncogenesis and predates liver metastasis, suggesting an earlier onset of metastasis in patients with PanNETs, providing novel insight into genetic changes in metastatic tumors of PanNETs.


Asunto(s)
Neoplasias Hepáticas , Neoplasias Primarias Secundarias , Tumores Neuroendocrinos , Neoplasias Pancreáticas , Variaciones en el Número de Copia de ADN/genética , Genómica , Humanos , Neoplasias Hepáticas/genética , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología
10.
Ann Transl Med ; 10(5): 262, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35402584

RESUMEN

Background: Cancer-associated fibroblasts (CAFs) are a vital constituent of the tumor microenvironment (TME) and have several activities, but the effect of CAF heterogeneity on the molecular features and clinical outcomes of pancreatic ductal adenocarcinoma (PDAC) remains unknown. Methods: An algorithm "scFrac" based on single-cell sequencing data from the Gene Expression Omnibus was introduced to emulate the enrichment of CAF subtypes in a TCGA-PDAC cohort and their prognostic influence, and confirmed by an external validation group (66 patients with PDAC) with multiplex immunohistochemistry staining. A comprehensive analysis including metabolic profile and transcription factor regulon activity was carried out among CAF subtypes. Results: Three distinct CAF populations were confirmed: myofibroblast (myCAF), inflammatory CAF (iCAF), and antigen-presenting CAF (apCAF). These subtypes expressed distinct metabolic profiles and transcriptional regulon activity. KEGG pathway annotation demonstrated that complement and coagulation cascades, as well as cytokine-cytokine receptor interaction were dominant in iCAFs, and pathways related to focal adhesion, and ECM-receptor interaction showed dominance in myCAFs, while antigen processing and presentation were the top enriched pathways in apCAFs. iCAFs trended to glycolysis with CREB3L1, EGR2 and SOX4 activation, whereas myCAFs depend on the tricarboxylic acid cycle and its derivatives with NRF2, CEBPD and YBX1 activation. iCAF is a protective factor associated with an inflammatory phenotype, but myCAF is an important factor in the poor prognosis of PDAC. Conclusions: We identified distinct molecular characteristics of 3 CAF subtypes in PDAC and plotted their metabolism profile. We introduced a novel algorism, scFrac, for exploring how CAF subgroups dysregulate cancer biology, and also shed a new therapeutic light on targeting the CAF subtype in TME.

11.
Oncogene ; 40(40): 5938-5949, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34363022

RESUMEN

Fructose-1,6-bisphosphatase (FBP1) is a rate-limiting enzyme in gluconeogenesis and an important tumor suppressor in human malignancies. Here, we aimed to investigate the expression profile of FBP1 in ovarian cancer, the molecular mechanisms that regulate FBP1 expression and to examine how the FBP1 regulatory axis contributes to tumorigenesis and progression in ovarian cancer. We showed that FBP1 expression was significantly decreased in ovarian cancer tissues compared with normal ovarian tissues, and low-FBP1 expression predicted poor prognosis in patients with ovarian cancer. The enhanced expression of FBP1 in ovarian cancer cell lines suppressed proliferation and 2-D/3-D invasion, reduced aerobic glycolysis, and sensitized cancer cells to cisplatin-induced apoptosis. Moreover, DNA methylation and C-MYC binding at the promoter inhibited FBP1 expression. Furthermore, through physical interactions with signal transducer and activator of transcription 3 (STAT3), FBP1 suppressed nuclear translocation of STAT3 and exerted its non-metabolic enzymatic activity to induce the dysfunction of STAT3. Thus, our study suggests that FBP1 may be a valuable prognostic predictor for ovarian cancer. C-MYC-dependent downregulation of FBP1 acted as a tumor suppressor via modulating STAT3, and the C-MYC/FBP1/STAT3 axis could be a therapeutic target.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Neoplasias Ováricas/genética , Factores de Transcripción/metabolismo , Animales , Proliferación Celular , Femenino , Fructosa-Bifosfatasa/metabolismo , Humanos , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Neoplasias Ováricas/patología , Factor de Transcripción STAT3 , Transducción de Señal , Transfección
12.
Cancer Commun (Lond) ; 41(6): 511-527, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33939321

RESUMEN

BACKGROUND: Although programmed cell death-ligand 1 (PD-L1) plays a well-known function in immune checkpoint response by interacting with programmed cell death-1 (PD-1), the cell-intrinsic role of PD-L1 in tumors is still unclear. Here, we explored the molecular regulatory mechanism of PD-L1 in the progression and metastasis of ovarian cancer. METHODS: Immunohistochemistry of benign tissues and ovarian cancer samples was performed, followed by migration, invasion, and angiogenesis assays in PD-L1-knockdown ovarian cancer cells. Immunoprecipitation, mass spectrometry, and chromatin immunoprecipitation were conducted along with zebrafish and mouse experiments to explore the specific functions and mechanisms of PD-L1 in ovarian cancer. RESULTS: Our results showed that PD-L1 induced angiogenesis, which further promoted cell migration and invasion in vitro and in vivo of ovarian cancer. Mechanistically, PD-L1 was identified to directly interact with vascular endothelial growth factor receptor-2 (VEGFR2) and then activated the FAK/AKT pathway, which further induced angiogenesis and tumor progression, leading to poor prognosis of ovarian cancer patients. Meanwhile, PD-L1 was found to be regulated by the oncogenic transcription factor c-JUN at the transcriptional level, which enhanced the expression of PD-L1 in ovarian cancer. Furthermore, we demonstrated that PD-L1 inhibitor durvalumab, combined with the antiangiogenic drug, apatinib, could enhance the effect of anti-angiogenesis and the inhibition of cell migration and invasion. CONCLUSION: Our results demonstrated that PD-L1 promoted the angiogenesis and metastasis of ovarian cancer by participating in the c-JUN/VEGFR2 signaling axis, suggesting that the combination of PD-L1 inhibitor and antiangiogenic drugs may be considered as a potential therapeutic approach for ovarian cancer patients.


Asunto(s)
Antígeno B7-H1 , Neoplasias Ováricas , Animales , Antígeno B7-H1/genética , Femenino , Humanos , Ratones , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Factor A de Crecimiento Endotelial Vascular , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Pez Cebra
13.
Cell Biol Toxicol ; 37(1): 35-49, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32399910

RESUMEN

Recepteur d'origine nantais (RON) has been implicated in cell proliferation, metastasis, and chemoresistance of various human malignancies. The short-form RON (sf-RON) encoded by RON transcripts was overexpressed in gastric cancer tissues, but its regulatory functions remain illustrated. Here, we found that sf-RON promoted gastric cancer cell proliferation by enhancing glucose metabolism. Furthermore, sf-RON was proved to induce the ß-catenin expression level through the AKT1/GSK3ß signaling pathway. Meanwhile, the binding sites of ß-catenin were identified in the promoter region of SIX1 and it was also demonstrated that ß-catenin positively regulated SIX1 expression. SIX1 enhanced the promoter activity of key proteins in glucose metabolism, such as GLUT1 and LDHA. Results indicated that sf-RON regulated the cell proliferation and glucose metabolism of gastric cancer by participating in a sf-RON/ß-catenin/SIX1 signaling axis and had significant implications for choosing the therapeutic target of gastric cancer.


Asunto(s)
Glucosa/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , beta Catenina/metabolismo , Animales , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Glucólisis , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Transducción de Señal , Análisis de Supervivencia
14.
Cell Oncol (Dordr) ; 42(5): 679-690, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31325096

RESUMEN

BACKGROUND: Apatinib is a tyrosine kinase inhibitor that targets vascular endothelial growth factor receptor-2 (VEGFR2), and has shown encouraging therapeutic effects in various malignant tumors. As yet, however, the role of apatinib in ovarian cancer has remained unknown. Here, we sought to elucidate the role of apatinib in the in vitro and in vivo viability and proliferation of ovarian cancer cells, as well as in glucose metabolism in these cells. METHODS: The effects of apatinib on ovarian cancer cell viability and proliferation were assessed using Cell Counting Kit-8 (CCK-8) and colony formation assays, respectively. The expression of VEGFR2/AKT1/SOX5/GLUT4 pathway proteins was assessed using Western blotting, and glucose uptake and lactate production assays were used to detect glycolysis in ovarian cancer cells. SOX5 was exogenously over-expressed and silenced in ovarian cancer cells using expression vector and shRNA-based methods, respectively. RNA expression analyses were performed using RNA-seq and gene-chip-based methods. GLUT4 promoter activity was assessed using a dual-luciferase reporter assay. The expression of p-VEGFR2 (Tyr1175), p-AKT1 (Ser473), p-GSK3ß (Ser9), SOX5 and GLUT4 in xenograft tissues was assessed using immunohistochemistry (IHC). RESULTS: We found that apatinib inhibited the in vitro and in vivo viability and proliferation in Hey and OVCA433 ovarian cancer cells in a dose-dependent and time-dependent manner. We also found that apatinib effectively suppressed glucose uptake and lactate production by blocking the expression of GLUT4 in these cells. In addition, we found that SOX5 predominantly rescued the inhibitory effect of apatinib on GLUT4 expression by activating its promoter. Finally, we found that apatinib regulated the expression of SOX5 by suppressing the VEGFR2/AKT1/GSK3ß signaling pathway. CONCLUSIONS: From our results, we conclude that apatinib suppresses the in vitro and in vivo viability and proliferation of ovarian cancer cells, as well as glycolysis by inhibiting the VEGFR2/AKT1/GSK3ß/SOX5/GLUT4 signaling pathway. Apatinib may serve as a promising drug for the treatment of ovarian cancer.


Asunto(s)
Antineoplásicos/farmacología , Glucólisis/efectos de los fármacos , Neoplasias Ováricas/metabolismo , Piridinas/farmacología , Factores de Transcripción SOXD/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Transportador de Glucosa de Tipo 4/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/uso terapéutico , Factores de Transcripción SOXD/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
15.
Theranostics ; 9(4): 1096-1114, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30867818

RESUMEN

The interplay between p53 and RAS signaling regulates cancer chemoresistance, but the detailed mechanism is unclear. In this study, we investigated the interactive effects of p53 and RAS on ovarian cancer cisplatin resistance to explore the potential therapeutic targets. Methods: An inducible p53 and RAS mutants active in either MAPK/ERK (S35 and E38) or PI3K/AKT (C40) or both (V12) were sequentially introduced into a p53-null ovarian cancer cell line-SKOV3. Comparative microarray analysis was performed using Gene Chip Prime View Human Gene Expression arrays (Affymetrix). In vitro assays of autophagy and apoptosis and in vivo animal experiments were performed by p53 induction and/or cisplatin treatment using the established cell lines. The correlation between HDAC4 and HIF-1α or CREBZF and the association of HDAC4, HIF-1α, CREBZF, ERK, AKT, and p53 mRNA levels with patient survival in 523 serous ovarian cancer cases from TCGA was assessed. Results: We show that p53 and RAS mutants differentially control cellular apoptosis and autophagy to inhibit or to promote chemoresistance through dysregulation of Bax, Bcl2, ATG3, and ATG12. ERK and AKT active RAS mutants are mutually suppressive to confer or to deprive cisplatin resistance. Further studies demonstrate that p53 induces HIF-1α degradation and HDAC4 cytoplasmic translocation and phosphorylation. S35, E38, and V12 but not C40 promote HDAC4 phosphorylation and its cytoplasmic translocation along with HIF-1α. Wild-type p53 expression in RAS mutant cells enhances HIF-1α turnover in ovarian and lung cancer cells. Autophagy and anti-apoptotic processes can be promoted by the overexpression and cytoplasmic translocation of HDAC4 and HIF1-α. Moreover, the phosphorylation and cytoplasmic translocation of HDAC4 activate the transcription factor CREBZF to promote ATG3 transcription. High HDAC4 or CREBZF expression predicted poor overall survival (OS) and/or progression-free survival (PFS) in ovarian cancer patients, whereas high HIF-1α expression was statistically correlated with poor or good OS depending on p53 status. Conclusion: HIF-1α and HDAC4 may mediate the interaction between p53 and RAS signaling to actively control ovarian cancer cisplatin resistance through dysregulation of apoptosis and autophagy. Targeting HDAC4, HIF-1α and CREBZF may be considered in treatment of ovarian cancer with p53 and RAS mutations.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Autofagia , Cisplatino/farmacología , Resistencia a Antineoplásicos , Histona Desacetilasas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Represoras/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Análisis por Micromatrices , Trasplante de Neoplasias , Neoplasias Ováricas/tratamiento farmacológico , Transducción de Señal , Trasplante Heterólogo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas ras/metabolismo
16.
Cancer Res ; 79(10): 2604-2618, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30808674

RESUMEN

Retinoic acid-related orphan receptor C (RORC) is a member of the nuclear orphan receptor family and performs critical regulatory functions in cell proliferation, metastasis, and chemoresistance in various types of malignant tumors. Here we showed that expression of RORC is lost in tumor tissues of bladder cancer patients. Enhanced expression of RORC suppressed cell proliferation and glucose metabolism and increased cisplatin-induced apoptosis in vitro and in vivo. RORC bound the promoter region of programmed death ligand-1 (PD-L1) and negatively regulated PD-L1 expression. PD-L1 directly interacted with integrin ß6 (ITGB6) and activated the ITGB6/FAK signaling pathway. RORC prevented the nuclear translocation of STAT3 via suppression of the PD-L1/ITGB6 signaling pathway, which further inhibited bladder cell proliferation and glucose metabolism and increased cisplatin-induced apoptosis. These findings reveal that RORC regulates bladder cancer cell proliferation, glucose metabolism, and chemoresistance by participating in the PD-L1/ITGB6/STAT3 signaling axis. Moreover, this new understanding of PD-L1 signaling may guide the selection of therapeutic targets to prevent tumor recurrence. SIGNIFICANCE: These findings suggest that RORC-mediated regulation of a PD-L1/ITGB6/FAK/STAT3 signaling axis in bladder cancer provides several potential therapeutic targets to prevent tumor progression.


Asunto(s)
Antígeno B7-H1/genética , Proliferación Celular/genética , Resistencia a Antineoplásicos/genética , Glucólisis/genética , Cadenas beta de Integrinas/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Factor de Transcripción STAT3/genética , Neoplasias de la Vejiga Urinaria/genética , Adulto , Anciano , Anciano de 80 o más Años , Proliferación Celular/efectos de los fármacos , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Glucólisis/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/patología
17.
Oncol Rep ; 41(3): 1998-2008, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30747219

RESUMEN

Gastric cancer (GC) is one of the most lethal diseases worldwide, but the mechanism of GC development remains elusive. In the present study, the roles of stanniocalcin­1 (STC1) in GC were investigated. It was demonstrated that overexpression of STC1 mRNA and protein were associated with poor survival of patients with GC. The expression of STC1 was enhanced in hypoxic GC cells and overexpression of STC1 facilitated cell proliferation in hypoxia but not in normoxia. Furthermore, STC1 promoted chemoresistance, migration and invasion in hypoxia. Upregulating the expression of STC1 enhanced the expression of B cell lymphoma (Bcl)­2, neural­cadherin and matrix metalloproteinase­2, whereas it reduced the levels of cytochrome c, cleaved­caspase­9, cleaved­caspase­3 and epithelial­cadherin. However, downregulation of STC1 altered the expression of these proteins in the opposite direction. Furthermore, disturbing the expression of Bcl­2 partly reversed the changes to these proteins and also the pro­proliferation, anti­apoptosis and pro­invasion potential of STC1. In vivo experiments indicated that enhanced expression of STC1 promoted tumor growth and metastasis in mice. Collectively, the results indicated that STC1 may serve an oncogenic role in hypoxic GC via dysregulating Bcl­2, indicating that STC1 may be a potential therapeutic target in the treatment of GC.


Asunto(s)
Movimiento Celular/genética , Proliferación Celular/genética , Resistencia a Antineoplásicos/genética , Glicoproteínas/genética , Hipoxia/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Neoplasias Gástricas/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Hipoxia/patología , Masculino , Metaloproteinasa 2 de la Matriz/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , ARN Mensajero/genética , Estómago/patología , Neoplasias Gástricas/patología , Regulación hacia Arriba/genética
18.
Anticancer Drugs ; 30(1): 19-26, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30540594

RESUMEN

Barasertib is a highly selective Aurora kinase B (AURKB) inhibitor and has been widely applied in a variety of cancer cells to investigate the regulatory function of AURKB. However, the effect of barasertib on glucose metabolism in gastric cancer (GC) remains illustrated. Here, barasertib was identified to effectively reduce glucose uptake and lactate production in GC cells in a dose-dependent and time-dependent manner. The expression levels of GLUT1, LDHA and HK2 were decreased by barasertib treatment of GC cells. Furthermore, we found that barasertib induced the expression of ribosomal protein S7 (RPS7), as a tumor suppressor, to regulate glucose metabolism. Silencing of RPS7 rescued the effects of barasertib on glucose metabolism in GC cells. Overexpression of RPS7 suppressed the promoter activity of C-Myc, which has been identified as an important regulator of glucose metabolism in cancer cells. The clinical data showed that the expression level of AURKB in GC patients' sera and tissues were positively correlated with those of C-Myc, GLUT1 and LDHA, but negatively with that of RPS7. Therefore, these findings provide new evidence that barasertib regulates GC cell glucose metabolism by inducing the RPS7/C-Myc signal pathway, and have important implications for the development of therapeutic approaches using AURKB as a target protein to prevent tumor recurrence.


Asunto(s)
Aurora Quinasa B/antagonistas & inhibidores , Glucosa/metabolismo , Organofosfatos/farmacología , Quinazolinas/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Aurora Quinasa B/biosíntesis , Estudios de Casos y Controles , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Ribosómicas/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo
19.
Ann Surg Oncol ; 26(2): 635-643, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30374923

RESUMEN

BACKGROUND: Tumor-infiltrating neutrophils (TINs) indicate poor prognosis for patients with pancreatic ductal adenocarcinoma (PDAC). Activated neutrophils can generate neutrophil extracellular traps (NETs). Little is known about the presence and prognostic significance of tumor-infiltrating NETs in PDAC. METHODS: This study enrolled 317 patients, in two independent sets (training and validation), who underwent curative pancreatectomy for PDAC in Shanghai Cancer Center. TINs and NETs were identified by immunohistochemical staining for CD15 and citrullinated histone H3, respectively. The relationship between clinicopathological features and outcomes was analyzed. Accuracy of prognostic prediction models was evaluated using concordance index (C-index) and Akaike information criterion (AIC). RESULTS: NETs were associated with OS (both, P < 0.001) and RFS (both, P < 0.001) in the training and validation sets. Tumor-infiltrating NETs predicted poor postsurgical survival of patients with PDAC. Moreover, multivariate analysis identified NETs and AJCC TNM stage as two independent prognostic factors for OS and RFS. Combination of NETs with the 8th edition TNM staging system (C-index, 0.6994 and 0.6669, respectively; AIC, 1067 and 1126, respectively) generated a novel model that improved the predictive accuracy for survival in both sets (C-index, 0.7254 and 0.7117, respectively; AIC, 1047 and 1102, respectively). The model combining presence of NETs with the 7th edition AJCC TNM staging system also had improved predictive accuracy. CONCLUSIONS: NETs were an independent prognostic factor in PDAC and incorporation of NETs along with the standard TNM stating system refined risk-stratification and predicted survival in PDAC with improved accuracy.


Asunto(s)
Carcinoma Ductal Pancreático/mortalidad , Trampas Extracelulares , Pancreatectomía/mortalidad , Neoplasias Pancreáticas/mortalidad , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/cirugía , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Pronóstico , Tasa de Supervivencia , Neoplasias Pancreáticas
20.
Angiogenesis ; 22(1): 15-36, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30168025

RESUMEN

Pancreatic cancer is one of the most lethal malignancies worldwide. Although the standard of care in pancreatic cancer has improved, prognoses for patients remain poor with a 5-year survival rate of < 5%. Angiogenesis, namely, the formation of new blood vessels from pre-existing vessels, is an important event in tumor growth and hematogenous metastasis. It is a dynamic and complex process involving multiple mechanisms and is regulated by various molecules. Inhibition of angiogenesis has been an established therapeutic strategy for many solid tumors. However, clinical outcomes are far from satisfying for pancreatic cancer patients receiving anti-angiogenic therapies. In this review, we summarize the current status of angiogenesis in pancreatic cancer research and explore the reasons for the poor efficacy of anti-angiogenic therapies, aiming to identify some potential therapeutic targets that may enhance the effectiveness of anti-angiogenic treatments.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neovascularización Patológica , Neoplasias Pancreáticas , Animales , Humanos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA